• Nenhum resultado encontrado

Functional and genetic analysis of alpha-synuclein and huntingtin in Drosophila melanogaster

N/A
N/A
Protected

Academic year: 2019

Share "Functional and genetic analysis of alpha-synuclein and huntingtin in Drosophila melanogaster"

Copied!
211
0
0

Texto

(1)

Gonçalo Manuel Poças

Dissertation presented to obtain the Ph.D degree in Biochemistry

Instituto de Tecnologia Química e Biológica António Xavier | Universidade Nova de Lisboa

Oeiras,

December, 2015

(2)
(3)
(4)

Gonçalo Manuel Poças

Dissertation presented to obtain the Ph.D degree in Biochemistry

Instituto de Tecnologia Química e Biológica António Xavier | Universidade Nova de Lisboa

Oeiras, December, 2015

(5)

ii

Title: Functional and genetic analysis of alpha-synuclein and huntingtin in

Drosophila melanogaster

Dissertation presented to obtain the Ph.D. degree in Biochemistry

Author: Gonçalo Manuel Poças

Cell Signaling in Drosophila Laboratory

Instituto de Tecnologia Química e Biológica

Universidade Nova de Lisboa

Av. da República

Estação Agronómica Nacional

2780-157 Oeiras

Portugal

Front cover picture: image obtained by confocal microscopy of a Drosophila

brain expressing alpha-synuclein (green) in the dopaminergic neurons marked

by a specific antibody for Tyrosine Hydroxylase (blue).

Copyright © 2015 by Gonçalo Manuel Poças

(6)

I declare that the work presented in this thesis, except where otherwise stated,

is based on my own research and it was supervised by Doctor Pedro Manuel

Domingos. The work was mainly performed in Instituto de

Tecnologia Química e Biológica, Universidade Nova de Lisboa.

I am grateful for the financial support provided by Fundação para a Ciência e

(7)
(8)
(9)
(10)

Acknowledgements ... xi

Summary ... xiii

Sumário ... xv

Thesis publications ... xvii

List of acronyms ... xviii

Chapter I ... 1

General Introduction 1.1.Human neurodegenerative diseases ... 3

1.2.Parkinson’s disease ... 8

1.2.1. Epidemiology, symptoms and general molecular features ... 8

1.2.2. α-Synuclein: a major player in Parkinson’s disease ... 14

1.2.3. The vulnerability of dopaminergic neurons in Parkinson’s disease ... 17

1.3.Huntington’s disease ... 21

1.3.1. Epidemiology, symptoms and general molecular features ... 21

1.3.2. Huntingtin: the monogenic cause of Huntington’s disease ... 22

1.3.3. Medium spiny neurons vulnerability in Huntington’s disease ... 25

1.4.Modeling Parkinson’s and Huntington’s diseases in Drosophila ... 27

1.5.Main goals ... 35

1.6. References ... 36

Chapter II ... 49

The subcellular localization and axonal transport of α-synuclein in a Drosophila model for Parkinson’s disease 2.1. Abstract ... 51

2.2. Introduction ... 52

2.3. Results 2.3.1. The wild-type form of α-syn accumulates in the synaptic terminals of Drosophila photoreceptors ... 56

2.3.2. The A30P mutant version of α-syn is mislocalized in the Drosophila photoreceptors ... 57

2.3.3. No differences were detected in the aggregation state of the wild-type and the A30P mutant version of α-syn ... 59

2.3.4. Identification of α-syn WT and A30P protein interactors by co-immunoprecipitation and mass spectrometry analysis ... 62

(11)

viii

Chapter III ... 93

The role of huntingtin N-terminal phosphorylation on its aggregation and toxicity in a Drosophila model for Huntington’s disease 3.1. Abstract ... 95

3.2. Introduction ... 96

3.3. Results 3.3.1. Cdc25 inhibitors prevent mutant huntingtin aggregation and regulate its phosphorylation ... 100

3.3.2. The genetic knockdown of Cdc25 in Drosophila dopaminergic neurons reduced the aggregation of mutant Htt ... 103

3.3.3. The phosphorylation of NT17 residues modulates the oligomerization and aggregation of mutant Htt in human H4 glioma cells and in Drosophila dopaminergic neurons ... 105

3.3.4. The dephosphorylation of NT17 residues induces motor dysfunction and life span decrease in Drosophila ... 109

3.4. Discussion ... 112

3.5. Material and Methods... 114

3.6. References ... 119

Chapter IV ... 125

Crosstalk between Parkinson’s and Huntington’s diseases: α-synuclein modifies mutant huntingtin aggregation and neurotoxicity in Drosophila 4.1. Abstract ... 127

4.2. Introduction ... 128

4.3. Results 4.3.1. Co-expression of mutant Htt and α-syn alters Htt aggregation pattern .. 130

4.3.2. Htt103Q-mCherry and α-syn-EGFP co-localize and co-aggregate in dopaminergic neurons and in photoreceptor ... 132

4.3.3. Htt103Q-mCherry and α-syn-EGFP are physically interacting ... 135

4.3.4. Co-expression of Htt103Q-mCherry and α-syn-EGFP produces premature and severe degeneration in the photoreceptors ... 137

4.3.5. Co-expression of Htt103Q-mCherry and α-syn-EGFP in the nervous system causes severe motor dysfunction and a decrease in life span ... 139

4.4. Discussion ... 142

(12)

Chapter V ... 153

Testing the potential therapeutic effect of mannosylglycerate in Drosophila models for Parkinson’s and Huntington’s diseases 5.1. Abstract ... 155

5.2. Introduction ... 156

5.3. Results 5.3.1. Expression of MGSD in Drosophila ... 160

5.3.2. The co-expression of MGSD reduced the ER stress levels in photoreceptors expressing ninaEG69D ... 162

5.3.3. MG was not detected in cellular extracts from Drosophila tissues expressing MGSD ... 164

5.4. Discussion ... 168

5.5. Material and Methods... 170

5.6. References ... 173

Chapter VI ... 177

Final discussion 6.1. Final discussion ... 178

(13)
(14)

I would like to express my sincere gratitude to everyone who directly or

indirectly helped me to complete this thesis.

To my supervisor, Pedro Domingos, for allowing me to join his group

in 2008 when I was a master’s student, introducing me to the wonderful world

of Drosophila. Thank you for teaching me all the basics of Drosophila

genetics and crucial tricks and for giving me the freedom of exploring it on

my own during my PhD studies. Thank you for supporting me in the critical

phases of my PhD, both at the scientific and personal levels.

To all the present and former members of the group “Cell Signaling in

Drosophila” at ITQB. I would like to thank in particular to Fátima Cairrão, for

guiding me in some of the molecular biology techniques used in the beginning

of my PhD, to Vania Rasheva, for the support with Drosophila genetics, to

Catarina Gaspar, for all the nice scientific discussions, friendship and

companionship and to Yolanda Afonso, for helping me in the last 6 months in

some of my PhD projects.

To my collaborators Federico Herrera and Tiago Outeiro, who strongly

contributed to make the work in this thesis possible. I am also grateful to

Maria Luísa Vasconcelos for being part of my PhD thesis committee, together

with Tiago Outeiro, and for all the good scientific input and also for providing

me some fly stocks which were extremely useful during my PhD studies.

I also acknowledge the Fundação para Ciência e a Tecnologia (FCT) of

Ministry for Education and Science of Portugal for the indispensable financial

support under my PhD grant SFRH/BD/61477/2009. To Instituto de

Tecnologia Química e Biológica (ITQB) from Universidade Nova de Lisboa

for giving me the opportunity of performing my PhD studies in a very good

and stimulating environment, at the technical and intellectual levels. To

Instituto Gulbenkian de Ciência (IGC) for allowing me to use its facilities,

(15)

xii

and Margarida Saramago for the support and funny moments spent together.

To all the people from the former Tiago Outeiro’s group UNCM at IMM,

especially to Joana Branco-Santos for collaborating with me in some of my

PhD projects, to Patricia Guerreiro for helping me with the mass spectrometry

experiments and to Leonor Fleming for all her support, nice scientific

discussions and friendship.

To Diego Hartmann and Marija Petkovic for the friendship and

companionship during this long journey. Thank you for all the lunches we had

together, for all the funny moments, good conversations, advises and for

supporting me throughout all the frustrations and less positive periods but also

for sharing in the joy of the successes. To Luís Miguel Oliveira, who even

from the other side of the Atlantic Ocean, finds a way of encourage and

support me.

Finally, to all my dear friends and family for the love and support, with

especial words to my mother and my father, who always supported and

believed in me in all stages of my life, to my grandmother Deolinda, to my

grandfather António Melo and to my brother. To my dear grandparents Maria

do Carmo and Carlos Poças, who must be very proud, wherever they are. To

Jaime for all his support and words full of positive energy. To Filipe Martins

and Alpio James Aquilina for their great friendship and companionship. To

Rui for the extremely good moments during this last year, making everything

look a bit easier.

Muito obrigado a todos!

“Learn from yesterday, live for today, hope for tomorrow.

The important thing is not to stop questioning.”

(16)

In this work we established new transgenic Drosophila models for

Parkinson’s (PD) and Huntington’s disease (HD), two incurable devastating

human neurodegenerative diseases, which strongly compromise the patients’

motor abilities. Our Drosophila models are based on the transgenic

overexpression of fluorescent-tagged versions of two human neuronal proteins

extensively associated with these neuropathologies, but whose exact biological function is still unknown: alpha-synuclein (α-syn) for PD and huntingtin (Htt) for HD.

Here, we investigated whether the subcellular localization of α-syn and the defective axonal transport of this protein is relevant to the development of

PD. Using a Drosophila model for PD based on the overexpression of

EGFP-tagged versions of the wild-type and the familiar A30P mutant form of human α-syn, we observed a differential subcellular localization of the two versions of α-syn in the photoreceptors: while wild-type α-syn was enriched in pre-synaptic terminals, α-syn A30P was distributed throughout the cytoplasm of the photoreceptors, both in cell bodies and axons. We have identified, by

immunoprecipitation and mass spectrometry, the specific neuronal proteins interacting with wild-type and A30P α-syn, and by knocking-down the genes of these proteins we identified three candidates as neuronal modulators of α

-syn’s axonal transport and subcellular localization: Tomosyn, Spaghetti

Squash, and Synaptotagmin 4.

We also studied the role of N-terminal phosphorylation of mutant Htt in

HD. We used our Drosophila model for HD, based on the overexpression of a

mCherry-tagged version of the N-terminal truncated form of mutant human

Htt, encoded by the exon 1 of Htt gene (Httex1). We analyzed the relative

contribution of the phosphorylation state of each phosphorylatable residue in

(17)

xiv

neurotoxicity, depending on the biological context. Our findings suggest that

single phosphorylation events at NT17 could be a more effective therapeutic

approach against HD.

Taking advantage of our newly established Drosophila models for PD

and HD, we also investigated the possible crosstalk between these two neuropathologies, by studying the interaction of α-syn and Htt, at the genetic and functional levels. We showed that α-syn and mutant Htt co-aggregate in vivo when co-expressed in Drosophila and produce a synergistic

age-dependent increase in neurotoxicity associated to a decline in motor function and life span. Our results suggest that the co-existence of α-syn and Htt in the same neuronal cells worsens aggregation-related neuropathologies,

accelerating the disease progression.

Finally, we wanted to test the therapeutic properties of

mannosylglycerate (MG) in our Drosophila models for PD and HD. The

biosynthesis of MG can be catalyzed by MG synthase (MGSD). We

successfully generated transgenic lines expressing MGSD, but we could not

detect the biosynthesis and accumulation of MG in Drosophila.

We hope this work will contribute for a better understanding of the

molecular and cellular bases of PD and HD and that our new Drosophila

models for these pathologies may constitute one more useful platform

(18)

Neste trabalho estabelecemos novos modelos transgénicos de

Drosophila para as doenças de Parkinson (DP) e de Huntington (DH), duas

doenças humanas neurodegenerativas devastadoras e incuráveis que afectam

significativamente o controlo motor dos doentes. Os nossos modelos de

Drosophila baseiam-se na sobrexpressão de duas proteínas neuronais

humanas associadas a estas neuropatologias e para as quais a função biológica é desconhecida: alpha-synuclein (α-syn) para a DP e huntingtin (Htt) para a DH.

Um dos nossos objectivos consistia em investigar se a localização sub-celular da α-syn e o anormal transporte axonal desta proteína são relevantes no desenvolvimento da DP. Utilizando um modelo de Drosophila para a DP, baseado na sobrexpressão das formas “wild-type” e mutante A30P da α-syn fundidas com a “tag” fluorescente EGFP, observámos que o fenótipo relativo à localização sub-celular da α-syn é distinto para as duas formas da α-syn: a forma “wild-type” localiza-se predominantemente nos terminais

pré-sinápticos, enquanto que a mutante A30P distribui-se por todo o citoplasma

dos fotoreceptores, tanto nos corpos celulares como nos axónios. Através de

imunoprecipitação e espectrometria de massa, foi possível identificar as

proteínas que interagem especificamente com as versões “wild-type” e

mutante da α-syn e através do “knocking-down” dos genes que codificam para

estas proteínas conseguimos identificar três candidatos a moduladores da localização sub-celular e do transporte axonal da α-syn: Tomosyn, Spaghetti Squash, and Synaptotagmin 4.

Também estudámos o papel da fosforilação da porção N-terninal da Htt

mutante na DH. Para isso, utilizámos o nosso modelo de Drosophila para a

DH, baseado na sobrexpressão de uma versão truncada da porção N-terminal

da Htt mutante, codificada pelo exão 1 do gene da Htt (Httex1). Assim, foi

(19)

xvi

que eventos únicos de fosforilação no NT17 e fosfatases especificas modulam

efectivamente os níveis de agregação e neurotoxicidade da Htt mutante, de

uma forma que é dependente do contexto biológico. Assim, os nossos

resultados sugerem que eventos únicos de fosforilação no NT17 poderão

constituir uma estratégia terapêutica mais eficaz contra a DH.

Tirando partido dos novos modelos de Drosophila para as DP e DH

estabelecidos neste trabalho, o possível “crosstalk” entre estas duas doenças

foi também investigado, através do estudo da interacção genética e funcional

da α-syn e Htt. Desta forma, demonstrámos que a α-syn e a Htt mutante,

quando co-expressas em Drosophila, co-agregam “in vivo” e aumentam, de

uma forma sinergística e dependente da idade, a neurotoxicidade, disfunções

motoras e mortalidade. Assim, os nossos resultados sugerem que a co-existência da α-syn e Htt nas mesmas células neuronais poderá exacerbar as neuropatologias relacionadas com a agregação de proteínas, podendo acelerar

a progressão destas doenças.

Por último, também pretendiamos testar as propriedadades terapêuticas

do manosilglicerato (MG) nos nossos modelos de Drosophila para as DP e

DH. A biosíntese do MG pode ser catalizada pela MG sintetase (MGSD). Foi

possível produzir linhas transgénicas a expressar MGSD, mas não

conseguimos detectar a biosíntese e acumulação de MG em Drosophila.

Esperamos que este trabalho possa contribuir para um melhor

conhecimento das bases moleculares e celulares das DP e DH e que os nossos

novos modelos de Drosophila para estas patologias constituam mais uma

(20)

α-Synuclein modifies mutant Huntingtin aggregation and neurotoxicity in Drosophila. Human Molecular Genetics, 2015, 24(7):1898-907. DOI:

10.1093/hmg/ddu606

Manuscripts in preparation

Branco-Santos J*, Poças GM*, Herrera F, Domingos PM, Giorgini F, and

Outeiro TF N-terminal phosphorylation modulates mutant huntingtin

aggregation and neurotoxicity. * Equal contribution.

(21)

xviii

AD – Alzheimer’s disease

BiFC – Bimolecular Fluorescence Complementation

CAG – Cytosine-adenine-guanine triplet repeats

CNS – Central nervous system

Co-IP – Co-immunoprecipitation

CS – Compatible solutes

DA – Dopaminergic

DMSO – Dimethylsulfoxide

EIF4G1 – Eukaryotic translation initiation factor 4-gamma 1

GAL4/UAS – GAL4-dependent upstream activating sequence

GFP – Green fluorescent protein

GO – Gene ontology

GWAS – Genome Wide Association Studies

HD – Huntington’s disease

Htt – Huntingtin

Httex1 – Exon 1 of Htt gene

LDH – Lactate dehydrogenase

Lrrk2 – Leucine-rich repeat kinase 2

MG – Mannosylglycerate

MGSD – Mannosylglycerate synthase

MSN – Medium spiny neurons

(22)

PD – Parkinson’s disease

PolyQ – Polyglutamine

PRR – Proline-rich region

PTMs – Post-translational modifications

RP – Retinitis pigmentosa

S13 – Serine 13

S16 – Serine 16

SNCA – alpha-synuclein gene (non A4 component of amyloid precursor)

SNpc – Substantia nigra pars compacta

T3 – Threonine 3

TLC – Thin layer chromatography

UAS – Upstream activating sequence

VPS35 – Vacuolar protein sorting 35 homolog

VTA – Ventral tegmental area

(23)
(24)
(25)
(26)

1.1.Human neurodegenerative diseases

Human neurodegenerative diseases are a heterogeneous group of

devastating age-dependent disorders for which there is no cure or effective

symptomatic treatments.

These neuropathologies are characterized by the selective and

progressive loss of specific neuronal cells. The clinical manifestations of

each disease are determined by the region of the brain that degenerates

(Table 1.1). Although most neurodegenerative disorders display an array

of neural symptoms, they may be grouped and categorized in two major

groups: movement disorders and dementias. Movement disorders are

mainly characterized by the loss of motor control, in the form of tremor,

chorea, akinesia, bradykinesia or ataxia, for example. Parkinson’s disease

(PD) and Huntington’s disease (HD) belong to this category, although they

frequently show cognitive deficits and psychiatric problems concomitant

to motor deficits [1]. Dementias are mainly characterized by a severe loss

of cognitive function, as it occurs in Alzheimer’s disease (AD),

fronto-temporal dementia or dementia with Lewy bodies (LB). However,

dementias can also be accompanied by motor symptoms [2].

(27)

4

The genes responsible for many of the existing neurodegenerative

diseases have been identified, and they can be genetically inherited in a

recessive or dominant manner, depending on the disease. However, a high

percentage of cases are sporadic, being their causes and the associated

risks unclear. This is especially true for PD and AD, which are the two

most frequent neurodegenerative disorders [4]. Approximately 95% of PD

cases appear without a family history or a specific mutation [5].

The misfolding, aggregation and accumulation of neuronal proteins

into protein inclusions constitutes a common hallmark to most human

neurodegenerative diseases (Fig. 1.1).

(28)

Protein aggregates are visible in cells and tissues with basic

microscopic techniques, and still constitute the main post-mortem

diagnostic tool for neurodegenerative disorders, including PD [6, 7].

However, protein aggregation is a dynamic process that involves

seeding/nucleation mechanisms starting with the formation of small

soluble oligomeric species. This intermediate state of aggregation has a

high tendency to become stabilized by the formation of an oligomeric β-sheet structure, which by incorporation of additional monomers gives rise to protofibrils and finally to cross-β amyloid-like fibrils. Therefore protein oligomers constitute the nucleus material from which protein

aggregates start forming and growing, giving rise to the typical protein

amyloid-like inclusions observed in AD, PD and HD [8-10]. The generic term “amyloid” is commonly used to refer to the cross-β structure of the aggregates with binding affinities to Congo red and thioflavin S dies.

If large protein aggregates are the toxic entities responsible for

neurodegeneration or a protective response against other toxic species is

still widely debatable. There are several reports supporting a neurotoxic

role for fibrillary aggregates, in which large aggregates containing

misfolded proteins may interfere with the normal function of other

neuronal proteins, by sequestering them in these aggregates.[11-16].

However, the hypothesis that the large protein aggregates may

constitute a cellular defense mechanism against more toxic smaller

aggregates, or monomers, dimers, oligomers and protofibrils, has lately

(29)

6

Figure 1.2. Schematic representation of the process involved in the formation of amyloid inclusions in human neurodegenerative diseases. Several factors (such as, mutations, environmental stress and aging) may contribute to the misfolding and aggregation of native soluble monomer proteins. These aggregated monomers can adopt abnormal conformational structures, generating different intermediate species of aggregates (dimers, oligomers and protofibrils). A variety of complex pathways eventually give rise to large amyloid plaques, enriched in β-sheet fibrils, a histopathological hallmark of most human neurodegenerative diseases. These large aggregates were originally considered neurotoxic but accumulating evidences suggest they may have a cytoprotective role. Adapted from [17].

For example, in AD the cognitive impairment is not correlated with the density of Aβ plaques [18, 19], being the concentration of soluble Aβ much strongly correlated with the severity of the disease [20-22].

Equivalently, in PD LB could have a protective role, since the neurons

where these inclusions are mostly found are healthier than adjacent

neurons with no inclusions [23]. Soluble oligomeric species are often able

to cross membranes, move between cell compartments and outside the cell,

interacting with several macromolecules and consequently interfering with

(30)

The role of protein aggregates in neuropathologies remains poorly

understood and more studies are necessary to clarify if these aggregates

and the correspondent molecular mechanisms associated to protein

(31)

8

1.2. Parkinson’s disease

1.2.1.Epidemiology, symptoms and general molecular features

PD was firstly described in 1817 by James Parkinson as the

“shaking palsy” (Figure 1.3). The most prominent clinical symptoms are

related to motor functions: tremor, bradykinesia (slowness of movement),

rigidity and postural instability. In later stages, cognitive and behavioral

functions may also be affected. Post-mortem analyses of PD brains

revealed that PD is the second most frequent human neurodegenerative

disease after AD, affecting approximately 10 million patients worldwide.

Figure 1.3. First scientific description of Parkinson’s disease (PD) by James Parkinson in “An essay on the shanking palsy, 1817” and the illustration of this pathology by William Richard Gowers published in “A manual of diseases of the nervous system, 1886”.

PD is primarily an age-related disease and the number of affected

individuals is expected to increase significantly in the next decades as

(32)

PD, and neurodegenerative disorders in general, constitutes an

enormous socio-economic burden. For example, the current cost of the

treatments for patients with PD in United States is higher than 14 billion

dollars per year [26].

(33)

10

The presence of intraneuronal inclusions in surviving neurons,

called LB, constitutes one of the hallmarks of post-mortem brain analysis

of patients suffering from PD (Fig. 1.5). In PD the region of the brain most

affected by degeneration are the dopaminergic (DA) neurons from the

substantia nigra pars compacta (SNpc).

Figure 1.5. Lewy body (LB) and other cytoplasmic inclusions (CI) enriched in α -synuclein (α-syn) within the neurons of the dopaminergic neurons (DA) from substantia nigra pars compacta (SNpc) constitute an hallmark in Parkinson’s disease (PD). Extracted from [28].

PD is mostly sporadic, with familial forms constituting

approximately 10% of affected individuals. Eleven genes were associated

(34)

Table 1.2. Genetic loci associated to Mendelian Parkinson’s disease (PD). Extracted from [29].

Four of these loci are causative of autosomal dominant PD: SNCA,

encoding α-synuclein (α-syn); LRRK2, encoding leucine-rich repeat kinase

2; VPS35, encoding Vacuolar protein sorting 35 homolog and EIF4G1,

Eukaryotic translation initiation factor 4-gamma 1.

SNCA gene was the first to be identified as being associated to PD

and encodes α-syn, a neuronal protein of unknown function. Because α-syn is the principal component of the LB, it is considered a major player in this neuropathology. Several mutations have been mapped to SNCA

locus, including missense mutations and genomic duplications or

triplications, which account for the second most common cause of

dominant PD.

Mutations in LRRK2 are the most common genetic cause of

dominant PD, accounting for 10% of all familial forms. Lrrk2 is a

cytosolic protein with two predicted enzymatic domains, one with GTPase

and another with kinase activity. Although the exact biological function is

unknown, it has been associated with neurite formation and growth,

(35)

[30-12

34]. More recently, mutant versions of VPS35 and EIF4G1 were linked to

autosomal PD. VPS35 is one of the components of the tripartite complex

retromer, involved in the endosomal-lysosomal trafficking. The mutant

forms of VPS35 may affect the development of DA neurons through the

Wnt pathway [35, 36] andabnormal iron accumulation in the brain by the

DMT1 pathway [37, 38]. EIF4G1 is involved in mRNA translation [39].

Although the familial forms of PD constitute a minority, the

identification of PD loci has enabled the study and characterization of

some of the molecular mechanisms of idiopathic PD. For example, mutant

forms of Parkin, Pink 1 and F-box only protein 7 have been implicated in

the dysregulation of normal mitophagy [40-43], and mutations in the genes

encoding Lrrk2 and VPS35 may disrupt normal processes of protein

degradation by the autophagy/lysosomal pathway, ultimately leading to the

accumulation of cytotoxic misfolded proteins and cell death [44-47].

Besides the classical linkage analysis, genome-wide association

studies (GWAS) allowed to identify risk loci associated to sporadic forms

of PD (Table 1.3). However, the specific roles of these genes in the

molecular basis of PD remain unknown.

(36)

Table 1.3. Risk loci identified by several genome-wide association studies

(GWAS) as being associated with sporadic Parkinson’s disease (PD). Adapted

(37)

14

1.2.2 α-Synuclein: a major player in Parkinson’s disease

The synuclein family is constituted by α-, β- and γ-syn, but only α-syn has been found to be associated to human neurodegenerative diseases. α-Syn is a small neuronal protein composed by 140 amino acids, and encoded by the human SNCA gene in the chromosome 4 (Figure 1.6).

Figure 1.6. α-Synuclein (α-syn) is a major player in Parkinson’s disease pathogenesis. (A) α-syn is encoded by SNCA gene, located on chromosome 4. (B)

(38)

Several mutations segregated with PD have been already mapped to

the SNCA locus. The missense mutation A53T was mapped in 1997 [50].

Subsequently two additional missense mutations were mapped: A30P and

E46K [51, 52]. These missense mutations are located in the N-terminal

domain of α-syn, involved in the binding to membranes, and induce an

increase in the propensity of this protein to misfold and form fibrillar aggregates enriched in β-sheet structure [53-55]. More recently, the H50Q [56] and G51D [57] missense mutations were also identified. The H50Q

mutation stabilizes α-syn fibrils, significantly increasing the aggregation

rate and the ability of this protein to form amyloid inclusions, while the

G51D mutation slowdowns α-syn aggregation, but both mutations

increased the toxicity [58-61]. Genomic duplication or triplication of the SNCA locus, which increases α-syn expression, also causes a form of autosomal dominant PD [62-64]. The SNCA dosage is inversely correlated

with the age of onset and directly correlated with the severity of the

disease [65-67]. Post-mortem analysis of sporadic PD midbrain tissues revealed that the total mRNA levels for α-syn were significantly increased, when compared to control brains, further highlighting the relevance of the

levels of expression of this protein in PD [68].

GWAS indicated that variations at the SNCA locus are also strongly

correlated with the sporadic forms of PD, corroborating the importance of α-syn in the etiology of this neuropathology (Fig. 1.7). In fact, hundreds of single nucleotide polymorphisms (SNPs) within the SNCA locus have

been shown to increase susceptibility to PD (based on PDgene database – http://www.pdgene.org). Moreover, polymorphisms in the promoter region of SNCA have been also linked to PD [69]. The expansion of the

polymorphic dinucleotide repeat REP1 increases α-syn expression, thus

(39)

16

SNCA may also affect the stability and the alternative splicing of α-syn mRNA transcripts. Indeed, at least 6 different transcripts of this gene exist.

Besides the transcript encoding the full-length protein (140 aa), 5 truncated

forms have been described. One of these truncated forms (112 aa) has been

associated to LB formation and neurotoxicity [72].

Figure 1.7. Variations at the SNCA locus, identified by genome-wide association

studies (GWAS), associated to the sporadic forms of Parkinson’s disease (PD).

(A) The REP1 dinucleotide repeat, in the promoter region, regulates the levels of

SNCA expression; Three single nucleotide polymorphisms (SNP) that may alter

transcripts’ splicing and stability, and that are highly associated with PD, are also

indicated (in green). (B) Four isoforms of SNCA generated by alternative splicing

and implicated in the formation of Lewy bodies (LB) and neurotoxicity. Adapted

(40)

1.2.3 The vulnerability of dopaminergic neurons in Parkinson’s

disease

The specificity of the neuronal subpopulations that are mostly

affected and degenerate in each neurodegenerative disease raises a

fundamental question: are the insults specifically induced in those neurons

or are there specific cellular and molecular characteristics of the affected

neuronal subpopulations that render them more vulnerable to the insults?

In the case of PD, the most affected neuronal population is the DA neurons

from SNpc that project their axons to the striatum (Figure 1.8).

Figure 1.8. The dopaminergic (DA) system and the nigrostriatal pathway. The DA neurons from substantia nigra pars compacta (SNpc), the most vulnerable neuronal population in Parkinson’s disease (PD), project their axons to the medium spiny neurons (MSN) from the striatum, constituting the nigrostriatal pathway, which plays a crucial role in the motor control.

The death of these neurons, and the consequent impairment of the

nigrostriatal DA pathway, is the cause of the motor symptoms observed in

PD. It is especially intriguing if we consider that from all the types of DA

(41)

18

neurons from ventral tegmental area (VTA), the DA neurons from SNpc

are the most vulnerable in PD.

Protein misfolding and aggregation, dysregulation of the ubiquitin

proteasome system, mitochondrial dysfunction and oxidative stress are not

exclusive of DA neurons, but are present in most areas of the PD brain

[73-75]. The difficulty in justifying the vulnerability of the DA neurons

from SNpc also persists when considering the familial forms of PD. The

expression of genes linked to this pathology is not specific and limited to

the DA neurons. For example, SNCA is ubiquitously expressed in the

central nervous system (CNS) at high levels, being also present in other

non-neuronal tissues [76].

The exposure to the neurotoxins 6-OHDA and MPTP specifically

induces neurodegeneration of the DA neurons from SNpc, and for this

reason they have been largely used as pharmacological models of PD. The

selectivity of these neurotoxins is explained by the fact that their

internalization into the cells is dependent on Sodium-dependent dopamine

transporter (DAT), which is only expressed in the DA neurons.

Importantly, DATis expressed at higher levels in SNpc than in the VTA

[77]. However, this explanation cannot be applied to other drugs that

induce PD-like pathology and symptoms, as is the case of the toxins

rotenone and paraquat, widely used as pesticide and herbicide,

respectively. Rotenone affects the mitochondrial function by inhibiting the

mitochondrial complex I, while paraquat induces oxidative stress, both

insults not being specifically targeted to the DA neurons from SNpc.

Bolan and Pissadaki [78] and by Brichta and Greengard [79] have

previously reviewed several particular determinants that put the SNpc DA

“on the edge” of the risk to degenerate in PD. Some of these

characteristics are related with the neuroanatomy of the nigrostriatal

(42)

are unmyelinated and extremely long, having a total cumulative length of

70 cm [80] and the estimated number of synapses established by these

axons is equally massive (200.000 – 400.000) [81, 82]. These unique

morphological features impose extreme bioenergetic demands, increasing

the metabolic needs of the SNpc DA neurons to maintain the membrane

potential, generate action potentials and enable synaptic transmission [83].

These exceptional energetic demands could make them especially

susceptible to insults of any sort, including environmental and genetic

factors [78].

Another important risk factor inherent to the DA neurons is the

oxidation of dopamine and the consequent generation of reactive oxygen

species [84]. Dopamine is a potentially dangerous neurotransmitter within

the cells, as it easily undergoes oxidation generating dopamine-derived

quinones and other toxic molecules (Figure 1.9). Furthermore, cytoplasmic accumulations of misfolded α-syn may affect the secretory pathway by blocking the normal ER-Golgi traffic and thus exacerbating the deleterious

effects induced by toxic molecules, namely dopamine-derived products

(43)

20

Figure 1.9. Schematic representation of the molecular mechanisms associated to the toxic properties of the dopamine-derived quinones (DAQ). Extracted from [86].

The investigation of the specific cellular and molecular determinants

of the differential susceptibility of the DA neurons from SNpc in PD has

proven to be very challenging but holds great potential for the discovery of

(44)

1.3.Huntington’s disease

1.3.1. Epidemiology, symptoms and general molecular features

HD is the most common genetically inherited neurodegenerative

disease, belonging to the group of polyglutamine (polyQ) pathologies and

affecting approximately 5 to 10 per 100.000 individuals in the Caucasian

population [87-89]. It is caused by a mutation in the first exon of the

unconventionally large IT-15 gene (67 exons), which encodes a protein

called huntingtin (Htt).

Mutant Htt misfolds and accumulates into amyloid-like

proteinaceous aggregates in the medium spiny neurons (MSN) from the

striatum, being the most common post-mortem histopathological feature

(Fig. 1.10).

Figure 1.10. Post-mortem analysis of patients’ brains with Huntington’s disease (HD) show intranuclear inclusions (INI) and cytoplasmic inclusions (CI) containing mutant Huntingtin (Htt) accumulated. Extracted from [28].

Neurons from the thalamus, hippocampus and cortex also

degenerate, accounting for the typical symptoms observed in patients:

involuntary movements (chorea), dementia and psychiatric problems

(depression, anxiety, irritability, sexual dysfunction and suicidal

(45)

22

1.3.2. Huntingtin: the monogenic cause of Huntington’s disease

Since 1993, when the Htt gene was mapped to the short arm of

chromosome 4, intense research has been conducted aiming to identify the

biological function of Htt, as well as its role in the HD pathogenesis.

The disease-associated mutation of Htt occurs in the N-terminal

region of Htt and consists of an abnormally high number of

cytosine-adenine-guanine (CAG) triplet repeats. The expanded CAG repeats are

translated to an extended stretch of glutamines commonly called polyQ

tract [90, 91]. Htt is an unusually large protein, constituted by 3144 amino

acids (approximately 350 kDa). However, the N-terminal portion of

mutant Htt (exon 1) harboring the polyQ tract (Fig. 1.11) is sufficient to

produce HD phenotypes in vivo [92-97]. For this reason, most of the model

systems established to study HD in vitro and in vivo are based on the

expression of mutant versions of the exon 1 of Htt gene (Httex1).

(46)

The polyQ tract of wild-type Htt contains between 6 and 29

glutamines, while mutant alleles associated to HD contain over 36

glutamines. The longest polyQ tract ever detected in an HD patient was

constituted by 130 glutamines [98]. Individuals carrying intermediate

alleles containing a range of 29-35 glutamines are healthy and totally

asymptomatic. However, due to the meiotic instability of the CAG repeats,

these individuals have a very high probability to transmit a pathological

polyQ expansion over 36 glutamines to their offspring, especially from the

paternal side [99, 100].

The number of glutamines, which is responsible for approximately

70% of the variance, is inversely correlated with the age of onset of the

disease, with longer polyQ tracts inducing an earlier and more severe onset

of the pathology [101]. The mean age of HD onset is 38 years, although it

may vary between the ages of 25 and 70 years. PolyQ tracts containing

over 55 glutamines (5% of all cases) produce Juvenile HD, where the onset

can start before the age of 20 years [102]. The mean duration of the disease

is 17-20 years and patients usually die from pneumonia or suicide [103].

Despite the efforts made to date, there is still no cure for HD and the

drugs available can only treat the symptoms. For the development of

effective treatments for HD, it is essential to understand the cellular and

molecular mechanisms underlying this pathology.

The exact biological function of Htt is unknown, but it was shown to

possess an indispensable anti-apoptotic role [104, 105]. There is also

evidence that Htt is essential for normal development, as Htt-null mutant

mice die as embryos at day 7.5 [104, 106, 107]. Surprisingly, Htt is

dispensable for Drosophila development, but it is crucial for normal

long-term mobility and survival in adult flies [108]. Htt interacts with a high

(47)

24

functions, such as endocytosis, neurotransmission, transcriptional

regulation, axonal transport and apoptosis [109-111]. Htt could act as

scaffold, responsible for bringing together its protein partners and for

coordinating the transfer of information among different subcellular

compartments, namely between the nucleus and cytoplasm [111]. Htt

could also act as a scaffold protein in selective autophagy by promoting

cargo recognition and autophagy initiation [112].

The expansion of the polyQ tract promotes conformational changes

in Htt that dramatically increase its propensity to misfold and aggregate.

Htt aggregation could affect the interaction with its neuronal partners and

disturb the normal function of Htt and Htt-interacting proteins. This

suggests the involvement of both toxic gain-of-function and

loss-of-function mechanisms in the development of HD, by disrupting these

(48)

1.3.3. Medium spiny neurons vulnerability in Huntington’s disease

Although Htt is ubiquitously expressed throughout nervous system

and other peripheral tissues (namely, testes, liver, heart and lungs) the

pathological effects are predominantly induced in specific neuronal

populations of the brain, especially the MSN neurons from the striatum

[113-118]. The fact that mutant Htt affects this particular neuronal

population points to the possible relevance of cell- or tissue-specific

factors in HD pathogenesis, beyond the expanded polyQ tract.

Brain-derived neurotrophic factor (BDNF) is a possible determinant

of the increased susceptibility of MSN neurons to neurodegeneration in

HD. BDNF is a neurotrophin necessary for the development, survival and

proper function of the striatal neurons. Deficient BDNF signaling in the

striatum, presumably caused by mutant Htt, has been pointed as strong

candidate to be a molecular determinant and modulator in HD

(49)

26

(50)

1.4. Modeling Parkinson’s and Huntington’s diseases in Drosophila

The establishment of animal models is one of the most useful

approaches to study the pathogenic mechanisms of human diseases at the

molecular, cellular, organic and functional level. Drosophila

melanogaster, commonly known as fruit fly, is a powerful genetic model

organism that has been used to study complex biological phenomena for

more than a century, including human neurodegenerative diseases [121].

The identification of genes associated to the familial forms of PD

and HD in the last decade allowed the establishment of animal models for

these pathologies, which are essential to investigate the early

pre-symptomatic stages of pathogenesis and to test new drug candidates.

One of the key factors that contributed to the great success of

Drosophila as a model organism to study human pathologies is the very

high degree of conservation with mammals, since approximately 75% of

disease-related loci in humans have at least one Drosophila homologue

[122]. Drosophila also offers a great number of potent genetic tools, it has

a very well-known anatomy and has short life cycle and life-span. Finally,

the central nervous system of invertebrates and vertebrates shares a

common evolutionary origin. Flies have a complex nervous system

capable of learning and coordination of intricate behaviors, and there is a

significant degree of genetic and functional conservation between the fly

central complex and the human basal ganglia, which is the region

primarily affected in PD and HD [123]. Such homology constitutes an

undeniable advantage to model and study these and other human

neurodegenerative diseases.

In order to model a human neurodegenerative disease in flies, it is

necessary to express in this organism the human genes associated with this

(51)

28

Drosophila is by using the binary GAL4-dependent upstream activating

sequence (GAL4/UAS) system [124] (Fig. 1.13).

Figure 1.13. The GAL4/UAS system, used for overexpression of proteins in

Drosophila, is a binary system which allows for the ectopic expression of genes of

interest in a specific tissue or cell type. Two transgenic fly lines are created: the UAS line, in which the gene of interest is placed downstream of a UAS (Upstream Activating Sequence) domain, where the yeast transcriptional activator GAL4 binds; and the GAL4 line (driver), which expresses Gal4 under the control of a tissue specific promoter. The gene encoded in the UAS line is only activated when this line is crossed with the GAL4 line. In our study we generated UAS lines

encoding for α-synuclein (α-syn) and huntingtin (Htt) and we used pan-neuronal, dopaminergic and photoreceptor drivers.

The gene of interest is subcloned into an UAS expression construct,

which is microinjected into fly embryos to establish transgenic lines. The

protein of interest is expressed in a targeted way by performing the genetic

cross of the UAS line with a Gal4 line that expresses the yeast

transcriptional co-activator GAL4 in a specific tissue or cell type.

(52)

the presence of GAL4 in cells and tissues. Many cell-type and

developmentally regulated GAL4 lines, commonly called “drivers”, are

readily available from Drosophila public stock centers (e.g. Bloomington

Drosophila Stock Center at Indiana University). So, the effect of

expressing a human transgene in many different tissues and at various

developmental stages can be assayed without creating many independent

transgenic lines. The GAL4/UAS system is especially useful when one

aims to study the mechanisms of toxicity of human genes linked to the disease which are absent in the Drosophila genome, as is the case of α-syn.

In the first Drosophila model of PD, transgenic flies expressing wild-type or two familial mutant forms (A30P and A53T) of human α-syn in the brain reproduced key features of PD, including LB-like inclusions

(Fig. 1.14), selective degeneration of DA neurons, and abnormalities in the

locomotor behavior [125]. Actually, Drosophila reproduced PD

phenotypes better than mice models of the disease.

(53)

30

However, in another report, missexpression of α-syn in the Drosophila CNS did not cause death of the DA neurons [127]. The

inconsistency of the results from these independent studies may result

from the use of different technical approaches. Therefore, there is the need

for more independent studies in order to clarify some of these

inconsistencies and to generate Drosophila models that consistently

reproduce the key phenotypes resembling PD conditions.

Concerning HD, Drosophila models show the essential features

associated to the pathology, such as progressive neurodegeneration [128],

motor deficits, protein inclusions in cytoplasm and nucleus (Figure 1.15)

and a correlation between polyglutamine repeat length, age of onset and

severity of the phenotypes [129].

(54)

In this work we have established new transgenic Drosophila models

for PD and HD, based on the overexpression of wild-type and mutant versions of α-syn and Htt (Fig. 1.16).

Figure 1.16. We generated UAS transgenic lines encoding fluorescent-tagged versions of two human proteins associated with Parkinson’s (PD) and

Huntington’s diseases (HD). For PD we generated constructs of human α

-synuclein (α-syn) fused to EGFP and we used a wild-type (WT) and a familiar mutant form (the missense mutation A30P) of this protein. For HD we generated mCherry-tagged versions of a wild-type form with 19 glutamines (19Q) and a mutant form containing 97 glutamines (97Q) of human huntingtin (Htt).

For the PD model, we used the missense mutation A30P, associated

to familial cases of the disease; and for the HD model, we used wild-type

and mutant forms of Htt, with 19Q and 97Q in the polyQ tract, respectively. These proteins were fused to fluorescent tags (EGFP for α-syn and mCherry for Htt), allowing microscopy and co-localization

studies in either “live” or fixed biological materials.

Taking advantage of the Gal4/UAS system, we were able to induce

targeted expression of these proteins in different neuronal tissues of

interest, such as in the whole nervous system, using the pan-neuronal

driver Elav-Gal4, in the eye retina, using the sGMR-Gal4 driver and in the

(55)

32

Figure 1.17. Targeted expression of EGFP-tagged α-synuclein using the Gal4/UAS system. Confocal microscopy images of adult flies expressing α -synuclein-EGFP in different tissues: (A) in the whole-brain, using the pan-neuronal driver Elav-Gal4; (B) in the photoreceptors, using the GMR-Gal4 driver; (C) in the dopaminergic (DA) neurons, using the TH-Gal4 driver.

Throughout our study, depending on the specific questions and on

the particular experiments to be done, we induced the targeted expression

of the proteins of interest in different tissues, such as the whole brain, the

eye photoreceptors or the DA neurons. The DA neurons constituted one of

our favorite neuronal populations to study PD and HD in our Drosophila

models, being the TH-Gal4 driver extensively used in our study. This

driver expresses Gal4 under the control of the promotor region of tyrosine

hydroxylase (TH) gene. TH is the enzyme that catalyzes the rate-limiting

step of dopamine biosynthesis and is specifically expressed in all DA cells.

The DA neurons, besides being the neuronal cell type directly

affected in PD, the axons of the DA neurons constitute the main input to

the MSN from the striatum, affected in HD. The DA system in mammals is

involved in the control of locomotor behavior, motivational states and

cognitive function, all of them impaired to different degrees in PD and

HD. Furthermore, the impairment of particular behaviors, such as the

initiation of voluntary actions in mammals has been correlated with the

malfunction of specific subpopulations of DA neurons, namely the ones

(56)

Drosophila DA system is well characterized by means of dopamine

and anti-Tyrosine Hydroxylase (TH) immunoreactivity, enabling the

characterization of several DA clusters which were named according to

their anatomical localization in the brain: paired posterior lateral 1 and 2

(PPL1 and PPL2); paired posterior medial 1 and 2 (PPM1/2); paired

posterior medial 3 (PPM3); paired anterior lateral (PAL), and paired

anterior medial (PAM). (Fig. 1.18) [132-134]. Similarly to mammals,

Drosophila DA system is also involved in the modulation and control of

locomotor behavior. Furthermore, specific subset of DA neurons are

especially vulnerable to neurodegeneration and to induce motor deficits in

the context of PD, which is equivalent to the situation observed in humans

[135, 136].

For these reasons, we consider Drosophila DA neurons a very

useful and adequate system to model and study human neurodegenerative

(57)

34

(58)

1.5. Main goals

Using our newly established Drosophila models, we were interested

in the study of three different particular aspects that we believe to be

relevant to the molecular pathogenesis of PD and HD:

 the molecular determinants of the subcellular localization of α-syn

in PD (Chapter II)

 the effect of N-terminal phosphorylation in the aggregation and

toxicity of Htt in vitro and in vivo (Chapter III)

 the possible crosstalk between PD and HD molecular mechanisms,

by studying the genetic and functional interaction between α-syn

and Htt (Chapter IV)

Additionally, we tried to identify new potential therapeutic

compounds for PD and HD, being particularly interested in the putative

therapeutic effect of mannosylglycerate (MG) in our Drosophila models of

neurodegeneration (Chapter V). MG is a compatible solute, i.e. a substance

compatible with the cellular metabolism that accumulates in the cytoplasm

to balance external osmotic pressure. These properties confer potential

neuroprotective effects to this compound in the context human

neurodegenerative diseases, as previously shown by our collaborators

using a yeast model for PD [137].

We believe this work will contribute to a better understanding of the

molecular and cellular pathophysiology mechanisms underlying PD and

(59)

36

1.6. References

1. Perry, R.J. and J.R. Hodges, Spectrum of memory dysfunction in degenerative disease. Curr Opin Neurol, 1996. 9(4): p. 281-5. 2. Tsolaki, M., et al., Extrapyramidal symptoms and signs in

Alzheimer's disease: prevalence and correlation with the first symptom. Am J Alzheimers Dis Other Demen, 2001. 16(5): p. 268-78.

3. Soto, C., Unfolding the role of protein misfolding in neurodegenerative diseases. Nat Rev Neurosci, 2003. 4(1): p. 49-60.

4. Nussbaum, R.L. and C.E. Ellis, Alzheimer's disease and Parkinson's disease. N Engl J Med, 2003. 348(14): p. 1356-64.

5. van der Vegt, J.P., et al., Imaging the impact of genes on Parkinson's disease. Neuroscience, 2009. 164(1): p. 191-204. 6. Hughes, A.J., et al., The accuracy of diagnosis of parkinsonian

syndromes in a specialist movement disorder service. Brain, 2002. 125(Pt 4): p. 861-70.

7. Goedert, M., et al., 100 years of Lewy pathology. Nat Rev Neurol, 2013. 9(1): p. 13-24.

8. Jarrett, J.T., E.P. Berger, and P.T. Lansbury, Jr., The C-terminus of the beta protein is critical in amyloidogenesis. Ann N Y Acad Sci, 1993. 695: p. 144-8.

9. Scherzinger, E., et al., Self-assembly of polyglutamine-containing huntingtin fragments into amyloid-like fibrils: implications for Huntington's disease pathology. Proc Natl Acad Sci U S A, 1999. 96(8): p. 4604-9.

10. Wood, S.J., et al., alpha-synuclein fibrillogenesis is nucleation-dependent. Implications for the pathogenesis of Parkinson's disease. J Biol Chem, 1999. 274(28): p. 19509-12.

11. Lorenzo, A. and B.A. Yankner, Beta-amyloid neurotoxicity requires fibril formation and is inhibited by congo red. Proc Natl Acad Sci U S A, 1994. 91(25): p. 12243-7.

12. Pike, C.J., et al., Neurodegeneration induced by beta-amyloid peptides in vitro: the role of peptide assembly state. J Neurosci, 1993. 13(4): p. 1676-87.

(60)

14. Kayed, R., et al., Conformation dependent monoclonal antibodies distinguish different replicating strains or conformers of prefibrillar Abeta oligomers. Mol Neurodegener, 2010. 5: p. 57. 15. Okada, T., et al., Formation of toxic fibrils of Alzheimer's amyloid

beta-protein-(1-40) by monosialoganglioside GM1, a neuronal membrane component. J Mol Biol, 2007. 371(2): p. 481-9.

16. Novitskaya, V., et al., Amyloid fibrils of mammalian prion protein are highly toxic to cultured cells and primary neurons. J Biol Chem, 2006. 281(19): p. 13828-36.

17. Lotz, G.P. and J. Legleiter, The role of amyloidogenic protein oligomerization in neurodegenerative disease. J Mol Med (Berl), 2013. 91(6): p. 653-64.

18. Terry, R.D., et al., Some morphometric aspects of the brain in senile dementia of the Alzheimer type. Ann Neurol, 1981. 10(2): p. 184-92.

19. Hashimura, T., T. Kimura, and T. Miyakawa, Morphological changes of blood vessels in the brain with Alzheimer's disease. Jpn J Psychiatry Neurol, 1991. 45(3): p. 661-5.

20. Lue, L.F., et al., Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer's disease. Am J Pathol, 1999. 155(3): p. 853-62.

21. McLean, C.A., et al., Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer's disease. Ann Neurol, 1999. 46(6): p. 860-6.

22. Naslund, J., et al., Correlation between elevated levels of amyloid beta-peptide in the brain and cognitive decline. JAMA, 2000. 283(12): p. 1571-7.

23. Tompkins, M.M. and W.D. Hill, Contribution of somal Lewy bodies to neuronal death. Brain Res, 1997. 775(1-2): p. 24-9.

24. Agorogiannis, E.I., et al., Protein misfolding in neurodegenerative diseases. Neuropathol Appl Neurobiol, 2004. 30(3): p. 215-24. 25. Taylor, J.P., J. Hardy, and K.H. Fischbeck, Toxic proteins in

neurodegenerative disease. Science, 2002. 296(5575): p. 1991-5. 26. Kowal, S.L., et al., The current and projected economic burden of

Parkinson's disease in the United States. Mov Disord, 2013. 28(3): p. 311-8.

27. Dorsey, E.R., et al., Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030.

(61)

38

28. Ross, C.A. and M.A. Poirier, Opinion: What is the role of protein aggregation in neurodegeneration? Nat Rev Mol Cell Biol, 2005. 6(11): p. 891-8.

29. Spatola, M. and C. Wider, Genetics of Parkinson's disease: the yield. Parkinsonism Relat Disord, 2014. 20 Suppl 1: p. S35-8. 30. Winner, B., et al., Adult neurogenesis and neurite outgrowth are

impaired in LRRK2 G2019S mice. Neurobiol Dis, 2011. 41(3): p. 706-16.

31. Dachsel, J.C., et al., A comparative study of Lrrk2 function in primary neuronal cultures. Parkinsonism Relat Disord, 2010. 16(10): p. 650-5.

32. Tong, Y., et al., Loss of leucine-rich repeat kinase 2 causes age-dependent bi-phasic alterations of the autophagy pathway. Mol Neurodegener, 2012. 7: p. 2.

33. Ren, Y., et al., Selective vulnerability of dopaminergic neurons to microtubule depolymerization. J Biol Chem, 2005. 280(40): p. 34105-12.

34. Piccoli, G., et al., LRRK2 controls synaptic vesicle storage and mobilization within the recycling pool. J Neurosci, 2011. 31(6): p. 2225-37.

35. Castelo-Branco, G., et al., Differential regulation of midbrain dopaminergic neuron development by 1, 3a, and Wnt-5a. Proc Natl Acad Sci U S A, 2003. 100(22): p. 12747-52.

36. Port, F., et al., Wingless secretion promotes and requires retromer-dependent cycling of Wntless. Nat Cell Biol, 2008. 10(2): p. 178-85.

37. Deng, H., K. Gao, and J. Jankovic, The VPS35 gene and Parkinson's disease. Mov Disord, 2013. 28(5): p. 569-75.

38. Tabuchi, M., et al., Retromer-mediated direct sorting is required for proper endosomal recycling of the mammalian iron transporter DMT1. J Cell Sci, 2010. 123(Pt 5): p. 756-66.

39. Schulte, E.C., et al., Variants in eukaryotic translation initiation factor 4G1 in sporadic Parkinson's disease. Neurogenetics, 2012. 13(3): p. 281-5.

40. Kane, L.A., et al., PINK1 phosphorylates ubiquitin to activate Parkin E3 ubiquitin ligase activity. J Cell Biol, 2014. 205(2): p. 143-53.

(62)

42. Koyano, F., et al., Ubiquitin is phosphorylated by PINK1 to activate parkin. Nature, 2014. 510(7503): p. 162-6.

43. Lazarou, M., et al., The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature, 2015. 524(7565): p. 309-14.

44. Orenstein, S.J., et al., Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci, 2013. 16(4): p. 394-406.

45. Manzoni, C., et al., Inhibition of LRRK2 kinase activity stimulates macroautophagy. Biochim Biophys Acta, 2013. 1833(12): p. 2900-10.

46. Alegre-Abarrategui, J., et al., LRRK2 regulates autophagic activity and localizes to specific membrane microdomains in a novel human genomic reporter cellular model. Hum Mol Genet, 2009. 18(21): p. 4022-34.

47. Beilina, A., et al., Unbiased screen for interactors of leucine-rich repeat kinase 2 supports a common pathway for sporadic and familial Parkinson disease. Proc Natl Acad Sci U S A, 2014. 111(7): p. 2626-31.

48. Bras, J., R. Guerreiro, and J. Hardy, SnapShot: Genetics of Parkinson's disease. Cell, 2015. 160(3): p. 570-570 e1.

49. Venda, L.L., et al., alpha-Synuclein and dopamine at the crossroads of Parkinson's disease. Trends Neurosci, 2010. 33(12): p. 559-68.

50. Polymeropoulos, M.H., et al., Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science, 1997. 276(5321): p. 2045-7.

51. Kruger, R., et al., Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson's disease. Nat Genet, 1998. 18(2): p. 106-8. 52. Zarranz, J.J., et al., The new mutation, E46K, of alpha-synuclein

causes Parkinson and Lewy body dementia. Ann Neurol, 2004. 55(2): p. 164-73.

53. Coskuner, O. and O. Wise-Scira, Structures and free energy landscapes of the A53T mutant-type alpha-synuclein protein and impact of A53T mutation on the structures of the wild-type alpha-synuclein protein with dynamics. ACS Chem Neurosci, 2013. 4(7): p. 1101-13.

(63)

40

55. Lemkau, L.R., et al., Mutant protein A30P alpha-synuclein adopts wild-type fibril structure, despite slower fibrillation kinetics. J Biol Chem, 2012. 287(14): p. 11526-32.

56. Appel-Cresswell, S., et al., Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson's disease. Mov Disord, 2013. 28(6): p. 811-3.

57. Kiely, A.P., et al., alpha-Synucleinopathy associated with G51D SNCA mutation: a link between Parkinson's disease and multiple system atrophy? Acta Neuropathol, 2013. 125(5): p. 753-69. 58. Rutherford, N.J., et al., Divergent effects of the H50Q and G51D

SNCA mutations on the aggregation of alpha-synuclein. J Neurochem, 2014. 131(6): p. 859-67.

59. Porcari, R., et al., The H50Q mutation induces a 10-fold decrease in the solubility of alpha-synuclein. J Biol Chem, 2015. 290(4): p. 2395-404.

60. Lesage, S., et al., G51D alpha-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann Neurol, 2013. 73(4): p. 459-71.

61. Fares, M.B., et al., The novel Parkinson's disease linked mutation G51D attenuates in vitro aggregation and membrane binding of alpha-synuclein, and enhances its secretion and nuclear localization in cells. Hum Mol Genet, 2014. 23(17): p. 4491-509. 62. Singleton, A.B., et al., alpha-Synuclein locus triplication causes

Parkinson's disease. Science, 2003. 302(5646): p. 841.

63. Muenter, M.D., et al., Hereditary form of parkinsonism--dementia.

Ann Neurol, 1998. 43(6): p. 768-81.

64. Konno, T., et al., Autosomal dominant Parkinson's disease caused by SNCA duplications. Parkinsonism Relat Disord, 2015.

65. Miller, D.W., et al., Alpha-synuclein in blood and brain from familial Parkinson disease with SNCA locus triplication. Neurology, 2004. 62(10): p. 1835-8.

66. Ibanez, P., et al., Causal relation between alpha-synuclein gene duplication and familial Parkinson's disease. Lancet, 2004. 364(9440): p. 1169-71.

67. Chartier-Harlin, M.C., et al., Alpha-synuclein locus duplication as a cause of familial Parkinson's disease. Lancet, 2004. 364(9440): p. 1167-9.

(64)

69. Pihlstrom, L. and M. Toft, Genetic variability in SNCA and Parkinson's disease. Neurogenetics, 2011. 12(4): p. 283-93.

70. Goldman, S.M., et al., Head injury, alpha-synuclein Rep1 and Parkinson's disease: a meta-analytic view of gene-environment interaction. Eur J Neurol, 2015. 22(7): p. e75.

71. Janeczek, P., et al., Reduced expression of alpha-synuclein in alcoholic brain: influence of SNCA-Rep1 genotype. Addict Biol, 2014. 19(3): p. 509-15.

72. Kalivendi, S.V., et al., Oxidants induce alternative splicing of alpha-synuclein: Implications for Parkinson's disease. Free Radic Biol Med, 2010. 48(3): p. 377-83.

73. Cookson, M.R., Parkinsonism due to mutations in PINK1, parkin, and DJ-1 and oxidative stress and mitochondrial pathways. Cold Spring Harb Perspect Med, 2012. 2(9): p. a009415.

74. Lee, F.J. and F. Liu, Genetic factors involved in the pathogenesis of Parkinson's disease. Brain Res Rev, 2008. 58(2): p. 354-64.

75. Shen, J. and M.R. Cookson, Mitochondria and dopamine: new insights into recessive parkinsonism. Neuron, 2004. 43(3): p. 301-4.

76. Ltic, S., et al., Alpha-synuclein is expressed in different tissues during human fetal development. J Mol Neurosci, 2004. 22(3): p. 199-204.

77. Lammel, S., et al., Unique properties of mesoprefrontal neurons within a dual mesocorticolimbic dopamine system. Neuron, 2008. 57(5): p. 760-73.

78. Bolam, J.P. and E.K. Pissadaki, Living on the edge with too many mouths to feed: why dopamine neurons die. Mov Disord, 2012. 27(12): p. 1478-83.

79. Brichta, L. and P. Greengard, Molecular determinants of selective dopaminergic vulnerability in Parkinson's disease: an update.

Front Neuroanat, 2014. 8: p. 152.

80. Matsuda, W., et al., Single nigrostriatal dopaminergic neurons form widely spread and highly dense axonal arborizations in the neostriatum. J Neurosci, 2009. 29(2): p. 444-53.

81. Oorschot, D.E., Total number of neurons in the neostriatal, pallidal, subthalamic, and substantia nigral nuclei of the rat basal ganglia: a stereological study using the cavalieri and optical disector methods. J Comp Neurol, 1996. 366(4): p. 580-99.

82. Arbuthnott, G.W. and J. Wickens, Space, time and dopamine.

Imagem

Table 1.1. Clinical, pathological and biochemical features of neurodegenerative  diseases
Figure 1.1. The formation of protein aggregates constitutes a common hallmark of  human neurodegenerative diseases
Figure 1.2. Schematic representation of the process involved in the formation of  amyloid inclusions in human neurodegenerative diseases
Table  1.3.  Risk loci identified by several genome-wide association studies  (GWAS) as being associated with sporadic Parkinson’s disease (PD)
+7

Referências

Documentos relacionados

(A) Macroautophagy is composed of fine-tuned machinery that ensures specific target recognition and cargo delivery to the lysosome; (B) Accumulated -syn increases mTor and

Para melhor explicitar os resultados foram construídas quadro categorias, sendo elas: Distinção entre espiritualidade / religiosidade; Benefícios que os cuidados com

ROBERTA AMARAL.. Crianças e adolescentes passam apenas a produzir pintura, desenho, gravura e modelagem, cujo caráter técnico não espelha mais as possibilidades do que esse

Furthermore compared to wildtype littermates, in olfactory bulb of α Syn A53T mice at 10 months old we detected a marked decrease of cholinergic neurons in mitral cell layer and

Considering the significant loss of nAChRs in PD brain with a - synuclein over expression, the neurotoxicity of a -synuclein to SNc dopaminergic neurons, the extensive distribution

In addition, our animal model of PD indicates that attachment of CL1 to a -syn enhanced its toxicity to dopaminergic neurons in an age- dependent manner and induced the formation

PD patients’ respiratory limitations do not provide proper breathing support for them to produce normal phrases and loudness variation; at the phonatory level, the vocal folds

In the graph showing the evolution of the relative abundance of each conformer family (Figure 4.4) it is noticeable the effect protein concentration has in it, more precisely,